1
|
Giovannetti A, Guarnieri R, Petrizzelli F, Lazzari S, Padalino G, Traversa A, Napoli A, Di Giorgio R, Pizzuti A, Parisi C, Mazza T, Barbato E, Caputo V. Small RNAs and tooth development: The role of microRNAs in tooth agenesis and impaction. J Dent Sci 2024; 19:2150-2156. [PMID: 39347023 PMCID: PMC11437305 DOI: 10.1016/j.jds.2024.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/11/2024] [Indexed: 10/01/2024] Open
Abstract
Background/purpose Tooth development, or odontogenesis, is a complex process in which several molecular pathways play a key role. Recently, microRNAs, a class of approximately 20-nucleotide small RNA molecules that regulate gene expression, have been implicated in the odontogenesis process. This study aimed to assess the role of miRNAs in odontogenesis anomalies, specifically agenesis and impaction. Materials and methods We analyzed a manually curated list of 82 miRNAs associated with human odontogenesis, sourced from literature data. Employing two different approaches to validate findings, we conducted functional enrichment analysis to evaluate the cell pathways, diseases, and phenotypes enriched for those miRNAs. Results Our findings indicate that the analyzed miRNAs regulate pathways linked to tooth anomalies, including the TGFꞵ and Wnt signaling pathways, and those governing the pluripotency of stem cells, known to mediate various cellular processes, and interconnected with odontogenesis-related pathways. Furthermore, the analysis disclosed several pathways associated with tumors, including small cell lung and gastric cancer. These results were confirmed also by diseases and phenotypes enrichment evaluation. Moreover, cell network analysis disclosed that miRNAs are embedded and interconnected in networks associated with dental diseases and cancer development, thus confirming the functional enrichment analyses. Conclusion In summary, our results offer a quantitative measure of the potential involvement of miRNAs in regulating pathways crucial for developmental processes, notably odontogenesis, and provide results suggesting potential association with oncogenesis processes as well.
Collapse
Affiliation(s)
- Agnese Giovannetti
- Clinical Genomics Laboratory, Fondazione IRCCS Casa Sollievo della Sofferenza, S. Giovanni Rotondo (FG), Italy
| | - Rosanna Guarnieri
- Department of Oral and Maxillofacial Sciences, School of Dentistry, Sapienza University of Rome, Rome, Italy
| | - Francesco Petrizzelli
- Bioinformatics Laboratory, Fondazione IRCCS Casa Sollievo della Sofferenza, S. Giovanni Rotondo (FG), Italy
| | - Sara Lazzari
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Gabriella Padalino
- Department of Oral and Maxillofacial Sciences, School of Dentistry, Sapienza University of Rome, Rome, Italy
| | - Alice Traversa
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
- Dipartimento di Scienze della Vita, della Salute e delle Professioni Sanitarie, Università degli Studi "Link Campus University", Roma, Italy
| | - Alessandro Napoli
- Bioinformatics Laboratory, Fondazione IRCCS Casa Sollievo della Sofferenza, S. Giovanni Rotondo (FG), Italy
| | | | - Antonio Pizzuti
- Clinical Genomics Laboratory, Fondazione IRCCS Casa Sollievo della Sofferenza, S. Giovanni Rotondo (FG), Italy
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Chiara Parisi
- Institute of Biochemistry and Cell Biology, CNR-National Research Council, Monterotondo Scalo, Rome, Italy
| | - Tommaso Mazza
- Bioinformatics Laboratory, Fondazione IRCCS Casa Sollievo della Sofferenza, S. Giovanni Rotondo (FG), Italy
| | - Ersilia Barbato
- Department of Oral and Maxillofacial Sciences, School of Dentistry, Sapienza University of Rome, Rome, Italy
| | - Viviana Caputo
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
2
|
Di Brina ALP, Palmieri O, Cannarozzi AL, Tavano F, Guerra M, Bossa F, Gentile M, Merla A, Biscaglia G, Cuttitta A, Perri F, Latiano A. Focus on Achalasia in the Omics Era. Int J Mol Sci 2024; 25:10148. [PMID: 39337632 PMCID: PMC11431880 DOI: 10.3390/ijms251810148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Achalasia is a rare and complex esophageal disease of unknown etiology characterized by difficulty in swallowing due to the lack of opening of the lower esophageal sphincter and the absence of esophageal peristalsis. Recent advancements in technology for analyzing DNA, RNA and biomolecules in high-throughput techniques are offering new opportunities to better understand the etiology and the pathogenetic mechanisms underlying achalasia. Through this narrative review of the scientific literature, we aim to provide a comprehensive assessment of the state-of-the-art knowledge on omics of achalasia, with particular attention to those considered relevant to the pathogenesis of the disease. The notion and importance of the multi-omics approach, its limitations and future directions are also introduced, and it is highlighted how the integration of single omics data will lead to new insights into the development of achalasia and offer clinical tools which will allow early diagnosis and better patient management.
Collapse
Affiliation(s)
- Anna Laura Pia Di Brina
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.L.P.D.B.); (O.P.); (A.L.C.); (F.T.); (M.G.); (F.B.); (M.G.); (A.M.); (G.B.); (F.P.)
| | - Orazio Palmieri
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.L.P.D.B.); (O.P.); (A.L.C.); (F.T.); (M.G.); (F.B.); (M.G.); (A.M.); (G.B.); (F.P.)
| | - Anna Lucia Cannarozzi
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.L.P.D.B.); (O.P.); (A.L.C.); (F.T.); (M.G.); (F.B.); (M.G.); (A.M.); (G.B.); (F.P.)
| | - Francesca Tavano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.L.P.D.B.); (O.P.); (A.L.C.); (F.T.); (M.G.); (F.B.); (M.G.); (A.M.); (G.B.); (F.P.)
| | - Maria Guerra
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.L.P.D.B.); (O.P.); (A.L.C.); (F.T.); (M.G.); (F.B.); (M.G.); (A.M.); (G.B.); (F.P.)
| | - Fabrizio Bossa
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.L.P.D.B.); (O.P.); (A.L.C.); (F.T.); (M.G.); (F.B.); (M.G.); (A.M.); (G.B.); (F.P.)
| | - Marco Gentile
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.L.P.D.B.); (O.P.); (A.L.C.); (F.T.); (M.G.); (F.B.); (M.G.); (A.M.); (G.B.); (F.P.)
| | - Antonio Merla
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.L.P.D.B.); (O.P.); (A.L.C.); (F.T.); (M.G.); (F.B.); (M.G.); (A.M.); (G.B.); (F.P.)
| | - Giuseppe Biscaglia
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.L.P.D.B.); (O.P.); (A.L.C.); (F.T.); (M.G.); (F.B.); (M.G.); (A.M.); (G.B.); (F.P.)
| | - Antonello Cuttitta
- Unit of Thoracic Surgery, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Francesco Perri
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.L.P.D.B.); (O.P.); (A.L.C.); (F.T.); (M.G.); (F.B.); (M.G.); (A.M.); (G.B.); (F.P.)
| | - Anna Latiano
- Division of Gastroenterology and Endoscopy, Fondazione IRCCS Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy; (A.L.P.D.B.); (O.P.); (A.L.C.); (F.T.); (M.G.); (F.B.); (M.G.); (A.M.); (G.B.); (F.P.)
| |
Collapse
|
3
|
Savarino EV, Salvador R, Ghisa M, Mari A, Forattini F, Costantini A, De Giorgio R, Zaninotto G. Research gap in esophageal achalasia: a narrative review. Dis Esophagus 2024; 37:doae024. [PMID: 38525929 DOI: 10.1093/dote/doae024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 03/08/2024] [Indexed: 03/26/2024]
Abstract
In recent years, new translational evidence, diagnostic techniques, and innovative therapies have shed new light on esophageal achalasia and revamped the attention on this relatively rare motility disorder. This narrative review aims to highlight the most recent progress and the areas where further research is needed. The four senior authors identified five topics commonly discussed in achalasia management: i.e. pathogenesis, role of functional lumen imaging probe in the diagnostic flow chart of achalasia, how to define the outcome of achalasia treatments, how to manage persistent chest pain after the treatment, and if achalasia patients' may benefit from a regular follow-up. We searched the bibliographic databases to identify systematic reviews, meta-analyses, randomized control trials, and original research articles in English up to December 2023. We provide a summary with the most recent findings in each of the five topics and the critical points where to address future research, such as the immune-genetic patterns of achalasia that might explain the transition among the different phenotypes, the need for a validated clinical definition of treatment success, the use of neuromodulators to manage chest pain, and the need for identifying achalasia patients at risk for cancer and who may benefit of long-term follow-up. Although undoubtedly, progress has been made on the definition and management of achalasia, unmet needs remain. Debated aspects range from mechanistic insights, symptoms, objective measure relationships, and accurate clinical responses to therapeutic interventions. Translational research is eagerly awaited to answer these unresolved questions.
Collapse
Affiliation(s)
- Edoardo Vincenzo Savarino
- Gastroenterology Unit, Azienda Ospedale Università of Padua, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Renato Salvador
- Chirurgia Generale 1, Azienda Ospedale Università of Padua, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Matteo Ghisa
- Gastroenterology Unit, Azienda Ospedale Università of Padua, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Amir Mari
- Gastroenterology Unit, Nazareth Hospital EMMS, The Azrieli Faculty of Medicine, Bar-Ilan University, Tel Aviv, Israel
| | - Francesca Forattini
- Chirurgia Generale 1, Azienda Ospedale Università of Padua, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Andrea Costantini
- Chirurgia Generale 1, Azienda Ospedale Università of Padua, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Roberto De Giorgio
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | | |
Collapse
|
4
|
Rodrigues DM, Lourenssen SR, Kataria J, Paterson WG, Blennerhassett MG, Bechara R. Altered Esophageal Smooth Muscle Phenotype in Achalasia. J Neurogastroenterol Motil 2024; 30:166-176. [PMID: 37528076 PMCID: PMC10999844 DOI: 10.5056/jnm23024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/12/2023] [Accepted: 05/11/2023] [Indexed: 08/03/2023] Open
Abstract
Background/Aims Achalasia is a disorder characterized by impairment in lower esophageal sphincter relaxation and esophageal aperistalsis, caused primarily by loss of inhibitory innervation. However, little is known about associated changes in esophageal smooth muscle. We examined the contractile phenotype and innervation of the circular smooth muscle, as well as inflammatory status, and correlated these with patient-specific parameters. Methods Circular smooth muscle biopsies were obtained in consecutive patients with achalasia undergoing peroral endoscopic myotomy. Axonal innervation and neurotransmitter subtypes were determined with immunocytochemistry, and this was used with quantitative Polymerase Chain Reaction (qPCR) to characterize smooth muscle proliferation and cellular phenotype, as well as collagen expression. These were compared to control tissue obtained at esophagectomy and correlated with patient demographic factors including age, onset of symptoms, and Eckhardt score. Results Biopsies of smooth muscle were obtained from 25 patients with achalasia. Overall, there was increased mast cell number and collagen deposition but increased smooth muscle cell proliferation vs control. There was a striking drop in axon density over controls, with no differences among subtypes of achalasia. Immunocytochemical analysis showed increased expression of the contractile marker α-smooth muscle actin, principally in Type 1 achalasia, that increased with disease duration, while qPCR identified increased mRNA for smoothelin with decreased myosin heavy chain and collagen 3a1, but not collagen 1a1. Conclusions The thickened circular smooth muscle layer in achalasia is largely denervated, with an altered contractile phenotype and fibrosis. Biopsies obtained during peroral endoscopic myotomy provide a means to further study the pathophysiology of achalasia.
Collapse
Affiliation(s)
- David M Rodrigues
- Division of Gastroenterology, Queen’s School of Medicine, Hotel Dieu Hospital, Kingston, ON, Canada
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Kingston, ON, Canada
| | - Sandra R Lourenssen
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Kingston, ON, Canada
| | - Jay Kataria
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Kingston, ON, Canada
| | - William G Paterson
- Division of Gastroenterology, Queen’s School of Medicine, Hotel Dieu Hospital, Kingston, ON, Canada
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Kingston, ON, Canada
| | - Michael G Blennerhassett
- Division of Gastroenterology, Queen’s School of Medicine, Hotel Dieu Hospital, Kingston, ON, Canada
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Kingston, ON, Canada
| | - Robert Bechara
- Division of Gastroenterology, Queen’s School of Medicine, Hotel Dieu Hospital, Kingston, ON, Canada
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Kingston, ON, Canada
| |
Collapse
|
5
|
Chanpong A, Alves MM, Bonora E, De Giorgio R, Thapar N. Evaluating the molecular and genetic mechanisms underlying gut motility disorders. Expert Rev Gastroenterol Hepatol 2023; 17:1301-1312. [PMID: 38117595 DOI: 10.1080/17474124.2023.2296558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 12/14/2023] [Indexed: 12/22/2023]
Abstract
INTRODUCTION Gastrointestinal (GI) motility disorders comprise a wide range of different diseases affecting the structural or functional integrity of the GI neuromusculature. Their clinical presentation and burden of disease depends on the predominant location and extent of gut involvement as well as the component of the gut neuromusculature affected. AREAS COVERED A comprehensive literature review was conducted using the PubMed and Medline databases to identify articles related to GI motility and functional disorders, published between 2016 and 2023. In this article, we highlight the current knowledge of molecular and genetic mechanisms underlying GI dysmotility, including disorders of gut-brain interaction, which involve both GI motor and sensory disturbance. EXPERT OPINION Although the pathophysiology and molecular mechanisms underlying many such disorders remain unclear, recent advances in the assessment of intestinal tissue samples, genetic testing with the application of 'omics' technologies and the use of animal models will provide better insights into disease pathogenesis as well as opportunities to improve therapy.
Collapse
Affiliation(s)
- Atchariya Chanpong
- Division of Gastroenterology and Hepatology, Department of Pediatrics, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
- Neurogastroenterology & Motility Unit, Gastroenterology Department, Great Ormond Street Hospital for Children, London, UK
| | - Maria M Alves
- Department of Clinical Genetics, Erasmus University Medical Center, Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Elena Bonora
- Department of Medical and Surgical Sciences, DIMEC, University of Bologna, Bologna, Italy
- U.O. Genetica Medica, IRCCS Azienda Ospedaliero-Universitaria di Bologna, AOUB, Bologna, Italy
| | - Roberto De Giorgio
- Department of Translational Sciences, University of Ferrara, Ferrara, Italy
| | - Nikhil Thapar
- Stem Cells and Regenerative Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
- Gastroenterology, Hepatology and Liver Transplant, Queensland Children's Hospital, Brisbane, Australia
- School of Medicine, University of Queensland, Brisbane, Australia
- Woolworths Centre for Child Nutrition Research, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
6
|
Chen S, Xing X, Hou X, Zhuang Q, Tan N, Cui Y, Wang J, Zhang M, Hu S, Xiao Y. The molecular pathogenesis of achalasia: a paired lower esophageal sphincter muscle and serum 4D label-free proteomic study. Gastroenterol Rep (Oxf) 2023; 11:goad031. [PMID: 37324545 PMCID: PMC10260389 DOI: 10.1093/gastro/goad031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/11/2023] [Accepted: 05/03/2023] [Indexed: 06/17/2023] Open
Abstract
Background Achalasia is a primary esophageal motility disorder with potential molecular pathogenesis remaining uncertain. This study aimed to identify the differentially expressed proteins and potential pathways among achalasia subtypes and controls to further reveal the molecular pathogenesis of achalasia. Methods Paired lower esophageal sphincter (LES) muscle and serum samples from 24 achalasia patients were collected. We also collected 10 normal serum samples from healthy controls and 10 normal LES muscle samples from esophageal cancer patients. The 4D label-free proteomic analysis was performed to identify the potential proteins and pathways involved in achalasia. Results Analysis of Similarities showed distinct proteomic patterns of serum and muscle samples between achalasia patients and controls (both P < 0.05). Functional enrichment analysis suggested that these differentially expressed proteins were immunity-, infection-, inflammation-, and neurodegeneration-associated. The mfuzz analysis in LES specimens showed that proteins involved in the extracellular matrix-receptor interaction increased sequentially between the control group, type III, type II, and type I achalasia. Only 26 proteins altered in the same directions in serum and muscle samples. Conclusions This first 4D label-free proteomic study of achalasia indicated that there were specific protein alterations in both the serum and muscle of achalasia, involving immunity, inflammation, infection, and neurodegeneration pathways. Distinct protein clusters between types I, II, and III revealed the potential molecular pathways associated with different disease stages. Analysis of proteins changed in both muscle and serum samples highlighted the importance of further studies on LES muscle and revealed potential autoantibodies.
Collapse
Affiliation(s)
| | | | - Xun Hou
- Gastrointestinal Surgery Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Qianjun Zhuang
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Niandi Tan
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Yi Cui
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Jinhui Wang
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Mengyu Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Shixian Hu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Yinglian Xiao
- Corresponding author. Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, Guangdong 510080, P. R. China. Tel: +86-13560172116;
| |
Collapse
|
7
|
Lu C, Wei F, He X, Yao X, Yu C. LncRNA expression in idiopathic achalasia: New insight and preliminary exploration into pathogenesis. Open Med (Wars) 2022; 17:732-740. [PMID: 35509690 PMCID: PMC9007103 DOI: 10.1515/med-2022-0473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 03/18/2022] [Accepted: 03/21/2022] [Indexed: 11/15/2022] Open
Abstract
Idiopathic achalasia is a primary esophageal motility disorder characterized by the absence of esophageal peristalsis and impaired relaxation of the lower esophageal sphincter (LES). However, the pathogenesis of idiopathic achalasia remains unclear. To further understand the pathogenesis, we conducted lncRNA and mRNA microarray analyses. LES specimens from 5 patients and 4 controls were used for microarray. Potential target genes with significantly changed lncRNA and mRNA were predicted using cis/trans-regulatory algorithms, followed by the Gene Ontology and KEGG pathway enrichment analysis to understand the biophysical effect. Finally, 7,133 significantly dysregulated mRNAs (3,136 increased and 3,997 decreased), along with 6,892 significantly dysregulated lncRNAs (4,900 increased and 1,992 decreased). Biophysical function analysis revealed that the cell adhesion molecule (CAM) pathway was a common pathway. The predicted lncRNA targets of NRXN1 (Down FC: 9.07), NTNG2 (UP FC: 2.75), CADM1 (Down FC: 2.26), NLGN1 (Down FC: 4.60), NEGR1 (Down FC: 2.335), CD22 (Down FC: 5.62), HLA-DQB1 (Down FC: 5.06), and HLA-DOA (Down FC: 2.31) were inputted in this pathway, which was mainly located in the synapse part of the neural system and immune system. Our study demonstrates the lncRNAs and corresponding mRNAs that may play important roles in idiopathic achalasia.
Collapse
Affiliation(s)
- Chao Lu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Furong Wei
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xinjue He
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xin Yao
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chaohui Yu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| |
Collapse
|
8
|
Yu XT, Wang F, Ding JT, Cai B, Xing JJ, Guo GH, Guo F. Tandem mass tag-based serum proteomic profiling revealed diabetic foot ulcer pathogenesis and potential therapeutic targets. Bioengineered 2022; 13:3171-3182. [PMID: 35068329 PMCID: PMC8974021 DOI: 10.1080/21655979.2022.2027173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Diabetic foot ulcer (DFU), one of the most serious complications of diabetes mellitus, is associated with a high amputation rate and decreased life quality. The impact of blood serum proteins on the occurrence and development of DFU has attracted a lot of interest. In this study, we aimed to define and compare the serum proteome of patients with DFU and healthy control (HC) to provide new insights into DFU pathogenesis. DFU patients and age- and sex-matched HCs were enrolled in this study (n = 54). We screened alterations in blood serum proteins from DFU patients and HC using a tandem mass tag (TMT) method based on liquid chromatography-mass spectrometry (LC-MS/MS) quantitative proteomics, and the differentially expressed proteins (DEPs) were further validated by parallel reaction monitoring (PRM) and enzyme-linked immunosorbent assay (ELISA). A total of 173 DEPs (100 up-regulated and 73 down-regulated) were identified between the DFU and HC groups (P < 0.05). Proteomic and bioinformatics analyses indicated that the proteins in the DFU group were mainly related to extracellular matrix (ECM)-receptor interaction and complement and coagulation cascades. The up-regulated DEPs were further verified by PRM and ELISA. LRG1, CD5L, CRP, IGHA1, and LBP were proved upregulated in DFU and these proteins are mainly related to immune response and complement activation. Our findings help to provide a more comprehensive understanding of the pathogenesis of DFU and new insight into potential therapeutic targets.
Collapse
Affiliation(s)
- Xiao-Ting Yu
- Burns Institute, the First Affiliated Hospital of Nanchang University, NanChang, JiangXi, China
| | - Feng Wang
- Ningbo Institute for Medicine & Biomedical Engineering Combined Innovation, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| | - Jia-Tong Ding
- Burns Institute, the First Affiliated Hospital of Nanchang University, NanChang, JiangXi, China
| | - Bo Cai
- Burns Institute, the First Affiliated Hospital of Nanchang University, NanChang, JiangXi, China
| | - Juan-Juan Xing
- Burns Institute, the First Affiliated Hospital of Nanchang University, NanChang, JiangXi, China
| | - Guang-Hua Guo
- Burns Institute, the First Affiliated Hospital of Nanchang University, NanChang, JiangXi, China
| | - Fei Guo
- Burns Institute, the First Affiliated Hospital of Nanchang University, NanChang, JiangXi, China
- Ningbo Institute for Medicine & Biomedical Engineering Combined Innovation, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
9
|
Panza A, Fontana A, Palmieri O, Merla A, Copetti M, Cuttitta A, Biscaglia G, Gentile A, Andriulli A, Latiano A. Circulating levels of cytokines, chemokines and growth factors in patients with achalasia. Biomed Rep 2021; 15:92. [PMID: 34631047 PMCID: PMC8461322 DOI: 10.3892/br.2021.1468] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 07/09/2021] [Indexed: 11/12/2022] Open
Abstract
Idiopathic achalasia is a disease that is characterized by the absence of peristalsis and incomplete relaxation of the lower esophageal sphincter, which is accompanied by dysphagia, regurgitation, chest pain and weight loss. The role of inflammatory infiltrates in the pathogenesis of achalasia remains controversial, although the infiltrating cell profile in the tissue has been previously characterized histologically and immunohistochemically. The present study aimed to evaluate the serum levels of 27 protein biomarkers to determine their association with achalasia and the clinical disease characteristics. The cytokine, chemokine and growth factor serum profiles of 68 patients with achalasia and 39 healthy individuals were explored using the 27-Bio-Plex Pro Human Cytokine assay. Reductions in the levels of inflammatory mediators IL-1β, IL-2, IL-5, IL-6, IL-8, IL-10, IL-12p70, IL-13, IL-15, IL-17, fibroblast growth factor, granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, interferon-γ, monocyte chemoattractant protein-1, macrophage inflammatory protein-1 (MIP-1)α and MIP-1β, regulated upon activation normal T cell expressed and presumably secreted, TNF-α and VEGF were detected in the serum samples of patients with achalasia compared with those in the control group (P<0.05). However, significant associations between the expression in the levels of inflammatory factors and clinical characteristics of the patients were not found (P>0.05). These results suggest that achalasia is a disease that has a local but not a systemic inflammatory pattern. Further studies are required to improve the current understanding of the mechanism underlying this disease.
Collapse
Affiliation(s)
- Anna Panza
- Fondazione IRCCS Casa Sollievo della Sofferenza, Gastroenterology Unit, I-71013 San Giovanni Rotondo, Foggia, Italy
| | - Andrea Fontana
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unit of Biostatistics, I-71013 San Giovanni Rotondo, Foggia, Italy
| | - Orazio Palmieri
- Fondazione IRCCS Casa Sollievo della Sofferenza, Gastroenterology Unit, I-71013 San Giovanni Rotondo, Foggia, Italy
| | - Antonio Merla
- Fondazione IRCCS Casa Sollievo della Sofferenza, Gastroenterology Unit, I-71013 San Giovanni Rotondo, Foggia, Italy
| | - Massimiliano Copetti
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unit of Biostatistics, I-71013 San Giovanni Rotondo, Foggia, Italy
| | - Antonello Cuttitta
- Fondazione IRCCS Casa Sollievo della Sofferenza, Unit of Thoracic Surgery, I-71013 San Giovanni Rotondo, Foggia, Italy
| | - Giuseppe Biscaglia
- Fondazione IRCCS Casa Sollievo della Sofferenza, Gastroenterology Unit, I-71013 San Giovanni Rotondo, Foggia, Italy
| | - Annamaria Gentile
- Fondazione IRCCS Casa Sollievo della Sofferenza, Gastroenterology Unit, I-71013 San Giovanni Rotondo, Foggia, Italy
| | - Angelo Andriulli
- Fondazione IRCCS Casa Sollievo della Sofferenza, Gastroenterology Unit, I-71013 San Giovanni Rotondo, Foggia, Italy
| | - Anna Latiano
- Fondazione IRCCS Casa Sollievo della Sofferenza, Gastroenterology Unit, I-71013 San Giovanni Rotondo, Foggia, Italy
| |
Collapse
|
10
|
GHOLIPOUR M, MIKAELI J, MOWLA SJ, BAKHTIARIZADEH MR, SAGHAEIAN JAZI M, JAVID N, FAZLOLLAHI N, KHOSHNIA M, BEHNAMPOUR N, MORADI A. Identification of differentially expressed microRNAs in primary esophageal achalasia by next-generation sequencing. Turk J Biol 2021; 45:262-274. [PMID: 34377051 PMCID: PMC8313935 DOI: 10.3906/biy-2101-61] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/08/2021] [Indexed: 11/29/2022] Open
Abstract
Molecular knowledge regarding the primary esophageal achalasia is essential for the early diagnosis and treatment of this neurodegenerative motility disorder. Therefore, there is a need to find the main microRNAs (miRNAs) contributing to the mechanisms of achalasia. This study was conducted to determine some patterns of deregulated miRNAs in achalasia. This case-control study was performed on 52 patients with achalasia and 50 nonachalasia controls. The miRNA expression profiling was conducted on the esophageal tissue samples using the next-generation sequencing (NGS). Differential expression of miRNAs was analyzed by the edgeR software. The selected dysregulated miRNAs were additionally confirmed using the quantitative reverse transcription polymerase chain reaction (qRT-PCR). Fifteen miRNAs were identified that were significantly altered in the tissues of the patients with achalasia. Among them, three miRNAs including miR-133a-5p, miR-143-3p, and miR-6507-5p were upregulated. Also, six miRNAs including miR-215-5p, miR-216a-5p, miR-216b-5p, miR-217, miR-7641 and miR-194-5p were downregulated significantly. The predicted targets for the dysregulated miRNAs showed significant disease-associated pathways like neuronal cell apoptosis, neuromuscular balance, nerve growth factor signaling, and immune response regulation. Further analysis using qRT-PCR showed significant down-regulation of hsa-miR-217 (p-value = 0.004) in achalasia tissue. Our results may serve as a basis for more future functional studies to investigate the role of candidate miRNAs in the etiology of achalasia and their application in the diagnosis and probably treatment of the disease.
Collapse
Affiliation(s)
- Mahin GHOLIPOUR
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, GorganIran
| | - Javad MIKAELI
- Autoimmune and Motility Disorders Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, TehranIran
| | - Seyed Javad MOWLA
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, TehranIran
| | | | - Marie SAGHAEIAN JAZI
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, GorganIran
| | - Naeme JAVID
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, GorganIran
| | - Narges FAZLOLLAHI
- Autoimmune and Motility Disorders Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, TehranIran
| | - Masoud KHOSHNIA
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, GorganIran
| | - Naser BEHNAMPOUR
- Department of Biostatistics, Faculty of Health, Golestan University of Medical Sciences, GorganIran
| | - Abdolvahab MORADI
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, GorganIran
- Department of Microbiology, Faculty of Medicine, Golestan University of Medical Sciences, GorganIran
| |
Collapse
|
11
|
Wu XY, Liu ZQ, Wang Y, Chen WF, Gao PT, Li QL, Zhou PH. The etiology of achalasia: An immune-dominant disease. J Dig Dis 2021; 22:126-135. [PMID: 33583137 DOI: 10.1111/1751-2980.12973] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023]
Abstract
There is accumulating evidence suggesting that an autoimmune component is involved in esophageal achalasia. An increase in immune cells, cytokines, chemokines, and autoimmune antibodies in serum and infiltration of immune cells in tissues support the view that immune-mediated inflammation is a crucial pathogenesis of inhibitory neuron degeneration in the lower esophageal sphincter. Infection of viruses such as the herpes virus family has been suspected of provoking the autoimmune reaction. Meanwhile, previous reports on immunogenetics have proposed that specific risk alleles on the human leukocyte antigen complex define the susceptible population to achalasia. In this study we reviewed current knowledge regarding the immune-related factors of achalasia, including immunology, viral infection and immunogenetic variations.
Collapse
Affiliation(s)
- Xing Yue Wu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Medical College, Fudan University, Shanghai, China
| | - Zu Qiang Liu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Wang
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Feng Chen
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ping Ting Gao
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Quan Lin Li
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ping Hong Zhou
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Qorri B, Tsay M, Agrawal A, Au R, Gracie J. Using machine intelligence to uncover Alzheimer’s disease progression heterogeneity. EXPLORATION OF MEDICINE 2020. [DOI: 10.37349/emed.2020.00026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Aim: Research suggests that Alzheimer’s disease (AD) is heterogeneous with numerous subtypes. Through a proprietary interactive ML system, several underlying biological mechanisms associated with AD pathology were uncovered. This paper is an introduction to emerging analytic efforts that can more precisely elucidate the heterogeneity of AD.
Methods: A public AD data set (GSE84422) consisting of transcriptomic data of postmortem brain samples from healthy controls (n = 121) and AD (n = 380) subjects was analyzed. Data were processed by an artificial intelligence platform designed to discover potential drug repurposing candidates, followed by an interactive augmented intelligence program.
Results: Using perspective analytics, six perspective classes were identified: Class I is defined by TUBB1, ASB4, and PDE5A; Class II by NRG2 and ZNF3; Class III by IGF1, ASB4, and GTSE1; Class IV is defined by cDNA FLJ39269, ITGA1, and CPM; Class V is defined by PDE5A, PSEN1, and NDUFS8; and Class VI is defined by DCAF17, cDNA FLJ75819, and SLC33A1. It is hypothesized that these classes represent biological mechanisms that may act alone or in any combination to manifest an Alzheimer’s pathology.
Conclusions: Using a limited transcriptomic public database, six different classes that drive AD were uncovered, supporting the premise that AD is a heterogeneously complex disorder. The perspective classes highlighted genetic pathways associated with vasculogenesis, cellular signaling and differentiation, metabolic function, mitochondrial function, nitric oxide, and metal ion metabolism. The interplay among these genetic factors reveals a more profound underlying complexity of AD that may be responsible for the confluence of several biological factors. These results are not exhaustive; instead, they demonstrate that even within a relatively small study sample, next-generation machine intelligence can uncover multiple genetically driven subtypes. The models and the underlying hypotheses generated using novel analytic methods may translate into potential treatment pathways.
Collapse
Affiliation(s)
- Bessi Qorri
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Mike Tsay
- NetraMark Corp, Toronto, ON M4E 1G8, Canada
| | | | - Rhoda Au
- Department of Anatomy & Neurobiology, Neurology and Epidemiology, Boston University Schools of Medicine and Public Health, Boston, MA 02218, USA
| | - Joseph Gracie
- NetraMark Corp, Toronto, ON M4E 1G8, Canada 5Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
13
|
Palmieri O, Mazza T, Bassotti G, Merla A, Tolone S, Biagini T, Cuttitta A, Bossa F, Martino G, Latiano T, Corritore G, Gioffreda D, Palumbo O, Carella M, Panza A, Andriulli A, Latiano A. microRNA-mRNA network model in patients with achalasia. Neurogastroenterol Motil 2020; 32:e13764. [PMID: 31773868 DOI: 10.1111/nmo.13764] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/07/2019] [Accepted: 10/28/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Achalasia is a rare idiopathic disease with a complex etio-pathogenesis still unknown. This study aimed to identify microRNA (miRNA)-mRNA regulatory networks underlying achalasia. METHODS The investigation was performed in tissue specimens from 11 patients and five controls using the microarray technology followed by an integrated bioinformatics analysis. KEY RESULTS One hundred and six miRNAs were significantly up-regulated and 64 were down-regulated in achalasia patients. The expression of the most 10 differential expressed miRNAs (miR-122-5p, miR-133a-3p, miR-504-5p, miR-187-3p, miR-133b, miR-200c-3p, miR-375, miR-200b-5p, miR-200b-3p, and miR203a) was confirmed by droplet digital PCR in an independent cohort. The interactions between the significant miRNAs and their targets uncovered 14 miRNA-mRNA interacting pairs with experimentally predicted genes (ie, FN1, ROCK2, DPYSL2), and 35 pairs with not experimentally target genes (ie, SULF1, MRVI1, PRKG1); all genes were involved in immune cell trafficking, skeletal and muscular system development, nervous system development macro-processes. CONCLUSION & INFERENCES The mRNA-miRNA regulatory networks described in this study provide new insights in the genetic background of the disease, suggesting further investigations in novel pathogenic mechanisms.
Collapse
Affiliation(s)
- Orazio Palmieri
- Gastroenterology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Gabrio Bassotti
- Department of Medicine, Gastroenterology and Hepatology Section, Santa Maria della Misericordia Hospital, Perugia, Italy
| | - Antonio Merla
- Gastroenterology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Salvatore Tolone
- Department of Surgery, University of Campania 'Luigi Vanvitelli', Napoli, Italy
| | - Tommaso Biagini
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Antonello Cuttitta
- Unit of Thoracic Surgery, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Fabrizio Bossa
- Gastroenterology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Giuseppina Martino
- Gastroenterology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Tiziana Latiano
- Gastroenterology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Giuseppe Corritore
- Gastroenterology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Domenica Gioffreda
- Gastroenterology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Orazio Palumbo
- Medical Genetics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Massimo Carella
- Medical Genetics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Anna Panza
- Gastroenterology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Angelo Andriulli
- Gastroenterology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Anna Latiano
- Gastroenterology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| |
Collapse
|
14
|
Albooshoke SN, Bakhtiarizadeh MR. Divergent gene expression through PI3K/akt signalling pathway cause different models of hypertrophy growth in chicken. ITALIAN JOURNAL OF ANIMAL SCIENCE 2019. [DOI: 10.1080/1828051x.2019.1634498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- S. N. Albooshoke
- Department of Animal Science, Khuzestan Agricultural and Natural Resources, Research and Education Center, AREEO, Ahwaz, Iran
| | - M. R. Bakhtiarizadeh
- Department of Animal Science, College of Aburaihan, Iran University of Tehran, Tehran, Iran
| |
Collapse
|
15
|
Nesteruk K, Spaander MCW, Leeuwenburgh I, Peppelenbosch MP, Fuhler GM. Achalasia and associated esophageal cancer risk: What lessons can we learn from the molecular analysis of Barrett's-associated adenocarcinoma? Biochim Biophys Acta Rev Cancer 2019; 1872:188291. [PMID: 31059738 DOI: 10.1016/j.bbcan.2019.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/02/2019] [Accepted: 04/29/2019] [Indexed: 02/08/2023]
Abstract
Idiopathic achalasia and Barrett's esophagus (BE) are preneoplastic conditions of the esophagus. BE increases the risk of esophageal adenocarcinoma (EAC), while achalasia is associated with both EAC and esophageal squamous cell carcinoma (ESCC). However, while the molecular mechanisms underlying the transformation of esophageal epithelial cells in BE are relatively well characterized, less is known regarding these processes in achalasia. Nevertheless, both conditions are associated with chronic inflammation and BE can occur in achalasia patients, and it is likely that similar processes underlie cancer risk in both diseases. The present review will discuss possible lessons that we can learn from the molecular analysis of BE for the study of achalasia-associated cancer and contrast findings in BE with those in achalasia. First, we will describe cellular fate during development of BE, EAC, and ESCC, and consider the inflammatory status of the epithelial barrier in BE and achalasia in terms of its contribution to carcinogenesis. Next, we will summarize current data on genetic alterations and molecular pathways involved in these processes. Lastly, the plausible role of the microbiota in achalasia-associated carcinogenesis and its contribution to abnormal lower esophageal sphincter (LES) functioning, the maintenance of chronic inflammatory status and influence on the esophageal mucosa through carcinogenic by-products, will be discussed.
Collapse
Affiliation(s)
- K Nesteruk
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M C W Spaander
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - I Leeuwenburgh
- Department of Gastroenterology and Hepatology, Franciscus Gasthuis, Rotterdam, the Netherlands
| | - M P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - G M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Rotterdam, the Netherlands..
| |
Collapse
|
16
|
Liu ZQ, Chen WF, Wang Y, Xu XY, Zeng YG, Lee Dillon D, Cheng J, Xu MD, Zhong YS, Zhang YQ, Yao LQ, Zhou PH, Li QL. Mast cell infiltration associated with loss of interstitial cells of Cajal and neuronal degeneration in achalasia. Neurogastroenterol Motil 2019; 31:e13565. [PMID: 30868687 DOI: 10.1111/nmo.13565] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/10/2018] [Accepted: 01/11/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Achalasia is a motility disorder of unknown etiology. Previous studies supported the hypothesis that autoimmune-mediated inflammatory responses produce inhibitory neuronal degeneration. This study was designed to explore the role of mast cells in achalasia. METHODS We collected information from 116 patients with achalasia who underwent peroral endoscopic myotomy between December 2016 and May 2017. Lower esophageal sphincter (LES) muscle biopsy was performed in all patients with achalasia, as well as 20 control subjects. The number of mast cells, interstitial cells of Cajal (ICCs), nNOS-positive cells, and S-100-positive cells in the LES were evaluated by immunohistochemistry. Pathological and clinical data were compared between groups. KEY RESULTS Compared with controls, the LES of patients with achalasia had significantly fewer ICCs, nNOS-positive cells, and S-100-positive cells and a higher number of mast cells (all P < 0.001). Furthermore, the increased mast cell infiltration was significantly associated with decreased ICCs, nNOS-positive cells, and S-100-positive cells in patients with achalasia (all P < 0.05). Clinically, the number of strongly positive mast cells was highest in patients with type I achalasia and lowest in those with type III achalasia (P < 0.001). In addition, patients with a history of autoimmune disease or viral infection had greater mast cell infiltration in the LES muscle (P = 0.040). CONCLUSIONS & INFERENCES In patients with achalasia, mast cell infiltration in the LES muscle is increased, in association with loss of ICCs and neuronal degeneration. Mast cells may thereby play a crucial role in the development of achalasia.
Collapse
Affiliation(s)
- Zu-Qiang Liu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei-Feng Chen
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Wang
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Yue Xu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi-Gang Zeng
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dustin Lee Dillon
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Jing Cheng
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mei-Dong Xu
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun-Shi Zhong
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi-Qun Zhang
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li-Qing Yao
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ping-Hong Zhou
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Quan-Lin Li
- Endoscopy Center and Endoscopy Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Clayton S, Cauble E, Kumar A, Patil N, Ledford D, Kolliputi N, Lopes-Virella MF, Castell D, Richter J. Plasma levels of TNF-α, IL-6, IFN-γ, IL-12, IL-17, IL-22, and IL-23 in achalasia, eosinophilic esophagitis (EoE), and gastroesophageal reflux disease (GERD). BMC Gastroenterol 2019; 19:28. [PMID: 30744559 PMCID: PMC6371504 DOI: 10.1186/s12876-019-0937-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 01/22/2019] [Indexed: 02/08/2023] Open
Abstract
An elevation of serum inflammatory biomarkers in achalasia patients compared with controls recently was demonstrated. It has not been determined whether the elevation of inflammatory cytokines is unique to achalasia or occurs with other diseases involving the esophagus. The primary aim of our study was to compare the differences in plasma immunological profiles (TNF- α receptor, IL-6, IFN-γ, IL-12, IL-17, IL-22, and IL-23) of patients with achalasia, eosinophilic esophagitis (EoE), and gastroesophageal reflux disease (GERD). A secondary aim of this study was to classify these same plasma cytokine profiles in the three achalasia subtypes. METHODS Plasma from 53 patients with achalasia, 22 with EoE, and 20 with GERD (symptoms plus esophagitis or + reflux study) were analyzed. EXCLUSION CRITERIA malignancy, autoimmune condition, immunodeficiency disorder, and treatment with steroids/immune modulating drugs. Cytokine levels were assayed via multiplex enzyme-linked immunosorbent assay (ELISA). RESULTS Our key finding revealed significant elevations in IL- 6 (p = 0.0158) in achalasia patients compared with EoE patients. Overall, plasma inflammatory biomarker patterns were not different in the three subtypes of achalasia. CONCLUSION There were no differences between the cytokine levels of any of the measured biomarkers between the achalasia and GERD groups suggesting that luminal stasis does increase biomarker levels for any of the cytokines examined in our study. While these results are an early first step towards clarifying some aspects of the pathogenesis of achalasia, they bring about many more questions that require further investigation and expansion. Further investigation with a larger cohort and a broader panel of biomarkers is needed.
Collapse
Affiliation(s)
- Steven Clayton
- Wake Forest Baptist Medical Center, Winston-Salem, NC USA
| | - Elliot Cauble
- University of South Florida, Division of Gastroenterology, Florida, USA
| | - Ambuj Kumar
- University of South Florida, Division of Gastroenterology, Florida, USA
| | - Nirav Patil
- Greenville Health System, Greenville, SC USA
| | - Dennis Ledford
- University of South Florida, Division of Allergy Immunology, Florida, USA
| | | | | | - Donald Castell
- Medical University of South Carolina, Division of Gastroenterology, Charleston, SC USA
| | - Joel Richter
- University of South Florida, Division of Gastroenterology, Florida, USA
| |
Collapse
|
18
|
Venuto S, Castellana S, Monti M, Appolloni I, Fusilli C, Fusco C, Pucci P, Malatesta P, Mazza T, Merla G, Micale L. TRIM8-driven transcriptomic profile of neural stem cells identified glioma-related nodal genes and pathways. Biochim Biophys Acta Gen Subj 2018; 1863:491-501. [PMID: 30528352 DOI: 10.1016/j.bbagen.2018.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 11/26/2018] [Accepted: 12/03/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND We recently reported TRIM8, encoding an E3 ubiquitin ligase, as a gene aberrantly expressed in glioblastoma whose expression suppresses cell growth and induces a significant reduction of clonogenic potential in glioblastoma cell lines. METHODS we provided novel insights on TRIM8 functions by profiling the transcriptome of TRIM8-expressing primary mouse embryonal neural stem cells by RNA-sequencing and bioinformatic analysis. Functional analysis including luciferase assay, western blot, PCR arrays, Real time quantitative PCR were performed to validate the transcriptomic data. RESULTS Our study identified enriched pathways related to the neurotransmission and to the central nervous system (CNS) functions, including axonal guidance, GABA receptor, Ephrin B, synaptic long-term potentiation/depression, and glutamate receptor signalling pathways. Finally, we provided additional evidence about the existence of a functional interactive crosstalk between TRIM8 and STAT3. CONCLUSIONS Our results substantiate the role of TRIM8 in the brain functions through the dysregulation of genes involved in different CNS-related pathways, including JAK-STAT. GENERAL SIGNIFICANCE This study provides novel insights on the physiological TRIM8 function by profiling for the first time the primary Neural Stem Cell over-expressing TRIM8 by using RNA-Sequencing methodology.
Collapse
Affiliation(s)
- Santina Venuto
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy; Experimental and Regenerative Medicine, University of Foggia, Via A. Gramsci, 89/91, 71122, Foggia, Italy.
| | - Stefano Castellana
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Maria Monti
- CEINGE Advanced Biotechnology, Department of Chemical Sciences, Federico II University, Via Gaetano Salvatore, 486, 80145, Napoli, Italy.
| | - Irene Appolloni
- U.O. Medicina Rigenerativa Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy
| | - Caterina Fusilli
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Carmela Fusco
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Piero Pucci
- CEINGE Advanced Biotechnology, Department of Chemical Sciences, Federico II University, Via Gaetano Salvatore, 486, 80145, Napoli, Italy.
| | - Paolo Malatesta
- U.O. Medicina Rigenerativa Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genova, Italy; Department of Experimental Medicine (DiMES), University of Genova, Via Leon Battista Alberti, 2, 16132 Genova, Italy.
| | - Tommaso Mazza
- Bioinformatics Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Giuseppe Merla
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| | - Lucia Micale
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, Viale Padre Pio, 71013, San Giovanni Rotondo, Foggia, Italy.
| |
Collapse
|
19
|
Bonora E, Bianco F, Stanzani A, Giancola F, Astolfi A, Indio V, Evangelisti C, Martelli AM, Boschetti E, Lugaresi M, Ioannou A, Torresan F, Stanghellini V, Clavenzani P, Seri M, Moonen A, Van Beek K, Wouters M, Boeckxstaens GE, Zaninotto G, Mattioli S, De Giorgio R. INPP4B overexpression and c-KIT downregulation in human achalasia. Neurogastroenterol Motil 2018; 30:e13346. [PMID: 29644781 DOI: 10.1111/nmo.13346] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/06/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Achalasia is a rare motility disorder characterized by myenteric neuron and interstitial cells of Cajal (ICC) abnormalities leading to deranged/absent peristalsis and lack of relaxation of the lower esophageal sphincter. The mechanisms contributing to neuronal and ICC changes in achalasia are only partially understood. Our goal was to identify novel molecular features occurring in patients with primary achalasia. METHODS Esophageal full-thickness biopsies from 42 (22 females; age range: 16-82 years) clinically, radiologically, and manometrically characterized patients with primary achalasia were examined and compared to those obtained from 10 subjects (controls) undergoing surgery for uncomplicated esophageal cancer (or upper stomach disorders). Tissue RNA extracted from biopsies of cases and controls was used for library preparation and sequencing. Data analysis was performed with the "edgeR" option of R-Bioconductor. Data were validated by real-time RT-PCR, western blotting and immunohistochemistry. KEY RESULTS Quantitative transcriptome evaluation and cluster analysis revealed 111 differentially expressed genes, with a P ≤ 10-3 . Nine genes with a P ≤ 10-4 were further validated. CYR61, CTGF, c-KIT, DUSP5, EGR1 were downregulated, whereas AKAP6 and INPP4B were upregulated in patients vs controls. Compared to controls, immunohistochemical analysis revealed a clear increase in INPP4B, whereas c-KIT immunolabeling resulted downregulated. As INPP4B regulates Akt pathway, we used western blot to show that phospho-Akt was significantly reduced in achalasia patients vs controls. CONCLUSIONS & INFERENCES The identification of altered gene expression, including INPP4B, a regulator of the Akt pathway, highlights novel signaling pathways involved in the neuronal and ICC changes underlying primary achalasia.
Collapse
Affiliation(s)
- E Bonora
- Department of Medical and Surgical Sciences, DIMEC, University of Bologna and St. Orsola-Malpighi Hospital, Bologna, Italy
| | - F Bianco
- Department of Medical and Surgical Sciences, DIMEC, University of Bologna and St. Orsola-Malpighi Hospital, Bologna, Italy.,Department of Medical and Veterinary Sciences, DIMEVET, University of Bologna, Bologna, Italy
| | - A Stanzani
- Department of Medical and Surgical Sciences, DIMEC, University of Bologna and St. Orsola-Malpighi Hospital, Bologna, Italy.,Department of Medical and Veterinary Sciences, DIMEVET, University of Bologna, Bologna, Italy
| | - F Giancola
- Department of Medical and Surgical Sciences, DIMEC, University of Bologna and St. Orsola-Malpighi Hospital, Bologna, Italy.,Department of Medical and Veterinary Sciences, DIMEVET, University of Bologna, Bologna, Italy.,Centro di Ricerca Biomedica Applicata, St.Orsola-Malpighi Hospital, Bologna, Italy
| | - A Astolfi
- Interdepartmental Center for Cancer Research "G. Prodi" (CIRC), University of Bologna, Bologna, Italy
| | - V Indio
- Interdepartmental Center for Cancer Research "G. Prodi" (CIRC), University of Bologna, Bologna, Italy
| | - C Evangelisti
- Department of Experimental Medicine, DIMES, University of Bologna, Bologna, Italy
| | - A M Martelli
- Department of Experimental Medicine, DIMES, University of Bologna, Bologna, Italy
| | - E Boschetti
- Department of Medical and Surgical Sciences, DIMEC, University of Bologna and St. Orsola-Malpighi Hospital, Bologna, Italy.,Centro di Ricerca Biomedica Applicata, St.Orsola-Malpighi Hospital, Bologna, Italy
| | - M Lugaresi
- Department of Medical and Surgical Sciences, DIMEC, University of Bologna and St. Orsola-Malpighi Hospital, Bologna, Italy
| | - A Ioannou
- Department of Medical and Surgical Sciences, DIMEC, University of Bologna and St. Orsola-Malpighi Hospital, Bologna, Italy
| | - F Torresan
- Department of Digestive System, St. Orsola-Malpighi Hospital, Bologna, Italy
| | - V Stanghellini
- Department of Medical and Surgical Sciences, DIMEC, University of Bologna and St. Orsola-Malpighi Hospital, Bologna, Italy
| | - P Clavenzani
- Department of Medical and Veterinary Sciences, DIMEVET, University of Bologna, Bologna, Italy
| | - M Seri
- Department of Medical and Surgical Sciences, DIMEC, University of Bologna and St. Orsola-Malpighi Hospital, Bologna, Italy
| | - A Moonen
- Translational Research in GastroIntestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, KU Leuven University, Leuven, Belgium
| | - K Van Beek
- Translational Research in GastroIntestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, KU Leuven University, Leuven, Belgium
| | - M Wouters
- Translational Research in GastroIntestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, KU Leuven University, Leuven, Belgium
| | - G E Boeckxstaens
- Translational Research in GastroIntestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, KU Leuven University, Leuven, Belgium
| | - G Zaninotto
- Division of Surgery, Imperial College London, London, UK
| | - S Mattioli
- Department of Medical and Surgical Sciences, DIMEC, University of Bologna and St. Orsola-Malpighi Hospital, Bologna, Italy
| | - R De Giorgio
- Department of Medical Sciences, Nuovo Arcispedale S.Anna at Cona (Ferrara), University of Ferrara, Ferrara, Italy
| |
Collapse
|
20
|
Jia X, Min L, Zhu S, Zhang S, Huang X. Loss of sonic hedgehog gene leads to muscle development disorder and megaesophagus in mice. FASEB J 2018; 32:5703-5715. [DOI: 10.1096/fj.201701581r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Xueting Jia
- Department of GastroenterologyNational Clinical Research Center for Digestive Diseases, Beijing Digestive Disease CenterBeijing Key Laboratory for Precancerous Lesion of Digestive Diseases
- Department of StomatologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| | - Li Min
- Department of GastroenterologyNational Clinical Research Center for Digestive Diseases, Beijing Digestive Disease CenterBeijing Key Laboratory for Precancerous Lesion of Digestive Diseases
| | - Shengtao Zhu
- Department of GastroenterologyNational Clinical Research Center for Digestive Diseases, Beijing Digestive Disease CenterBeijing Key Laboratory for Precancerous Lesion of Digestive Diseases
| | - Shutian Zhang
- Department of GastroenterologyNational Clinical Research Center for Digestive Diseases, Beijing Digestive Disease CenterBeijing Key Laboratory for Precancerous Lesion of Digestive Diseases
| | - Xiaofeng Huang
- Department of StomatologyBeijing Friendship Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
21
|
Sato H, Takahashi K, Mizuno KI, Hashimoto S, Yokoyama J, Hasegawa G, Terai S. Esophageal motility disorders: new perspectives from high-resolution manometry and histopathology. J Gastroenterol 2018; 53:484-493. [PMID: 29134329 DOI: 10.1007/s00535-017-1413-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/09/2017] [Indexed: 02/04/2023]
Abstract
High-resolution manometry (HRM) and peroral endoscopic myotomy (POEM) have contributed significantly to the field of esophageal motility disorders in recent years. The development of HRM has categorized various esophageal motility disorders with a focus on a diverse range of manometric anomalies. Additionally, the Chicago classification criteria is widely used for manometric diagnosis. Moreover, POEM was introduced as a minimally invasive radical therapy for achalasia and shows promise for other spastic esophageal motility disorders as well. POEM has also enabled a transluminal endoscopic approach for determining the histology of the esophageal muscle layer, which is expected to assist in elucidating the etiology of disorders associated with esophageal motility. The purpose of this review is to update the diagnosis, pathology, and treatment of esophageal motility disorders, with a focus on the recent advances in this field.
Collapse
Affiliation(s)
- Hiroki Sato
- Division of Gastroenterology and Hepatology, Niigata University Medical and Dental Hospital, 757-1, Asahimachidori, Chuo-ku, Niigata, Niigata, 951-8510, Japan.
| | - Kazuya Takahashi
- Division of Gastroenterology and Hepatology, Niigata University Medical and Dental Hospital, 757-1, Asahimachidori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
| | - Ken-Ichi Mizuno
- Division of Gastroenterology and Hepatology, Niigata University Medical and Dental Hospital, 757-1, Asahimachidori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
| | - Satoru Hashimoto
- Division of Gastroenterology and Hepatology, Niigata University Medical and Dental Hospital, 757-1, Asahimachidori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
| | - Junji Yokoyama
- Division of Gastroenterology and Hepatology, Niigata University Medical and Dental Hospital, 757-1, Asahimachidori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
| | - Go Hasegawa
- Division of Cellular and Molecular Pathology, Department of Cellular Function, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Niigata University Medical and Dental Hospital, 757-1, Asahimachidori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
| |
Collapse
|
22
|
Peroral Endoscopic Myotomy in Children With Achalasia: A Relatively Long-term Single-center Study. J Pediatr Gastroenterol Nutr 2018; 66:257-262. [PMID: 28691974 DOI: 10.1097/mpg.0000000000001675] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Achalasia is a disease caused by neuromuscular dysfunction in the esophagogastric junction, with a relatively low incidence among children. Peroral endoscopic myotomy (POEM) is a new endoscopic procedure for the treatment of achalasia; however, it is rarely applied in children. OBJECTIVES We aim to study the feasibility, safety, and effectiveness of POEM as a treatment for pediatric achalasia. METHODS A total of 21 pediatric patients (ages from 11 months to 18 years) diagnosed as having achalasia and treated with POEM from October of 2014 to October of 2016 in our hospital were included in our study. Indices such as clinical manifestations, nutritional status, Eckardt scores, high-resolution esophageal manometric measurements, gastroduodenoscopic findings, and upper gastroenterographic findings after treatment were respectively compared with those before treatment to analyze the effectiveness and safety of POEM. RESULTS All of the 21 children successfully received POEM. The patients were followed up for duration ranged from 3 to 24 months. Among these children, symptoms such as vomiting and dysphagia were significantly alleviated or resolved. The Eckardt scores had an average drop of 7 points after treatment compared with those before treatment. Postoperative gastroduodenoscopy and upper gastroenterography showed that the opening of the cardiac orifice was significantly enlarged and obstruction at the lower esophagus was significantly relieved. CONCLUSIONS POEM is a mini-invasive technique and a promising new treatment for pediatric patients, making it worthy of further clinical research and application.
Collapse
|
23
|
Lei X, Muscat JE, Zhang B, Sha X, Xiu G. Differentially DNA methylation changes induced in vitro by traffic-derived nanoparticulate matter. Toxicology 2018; 395:54-62. [DOI: 10.1016/j.tox.2017.11.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 10/16/2017] [Accepted: 11/02/2017] [Indexed: 12/12/2022]
|